You are on page 1of 12

Critical Review

Regulation of Apoptosis by Heat Shock Proteins Donna Kennedy


1
Richard J ager
2
Dick D. Mosser
3
Afshin Samali
1
*
1
Department of Biochemistry, Apoptosis Research Centre, Biosciences
Research Building, Corrib Village, NUI Galway, Dangan, Galway, Ireland
2
Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied
Sciences, Rheinbach, Germany
3
Department of Molecular and Cellular Biology, University of Guelph,
Guelph, ON, Canada
Abstract
Thermotolerance, the acquired resistance of cells to stress, is
a well-established phenomenon. Studies of the key mediators
of this response, the heat shock proteins (HSPs), have led to
the discovery of the important roles played by these proteins
in the regulation of apoptotic cell death. Apoptosis is critical
for normal tissue homeostasis and is involved in diverse proc-
esses including development and immune clearance. Apopto-
sis is tightly regulated by both proapoptotic and antiapoptotic
factors, and dysregulation of apoptosis plays a signicant role
in the pathophysiology of many diseases. In the recent years,
HSPs have been identied as key determinants of cell survival,
which can modulate apoptosis by directly interacting with
components of the apoptotic machinery. Therefore, manipula-
tion of the HSPs could represent a viable strategy for the treat-
ment of diseases. Here, we review the current knowledge with
regard to the mechanisms of HSP-mediated regulation of apo-
ptosis. VC
2014 IUBMB Life, 66(5):327338, 2014
Keywords: cell death; apoptosis; intrinsic pathway; heat shock
proteins; caspases; heat shock response
Introduction
Heat shock proteins (HSPs) are an evolutionarily conserved
superfamily (1). As their name suggests, HSPs were originally
discovered to be upregulated after exposure of cells to elevated
temperatures; however, they are also induced in response to a
variety of other stress stimuli (2), and the heat shock response
is now recognized as being one of the most ancient and evolu-
tionarily conserved cytoprotective mechanisms found in nature
(3). Subjecting cells to a bout of mild thermal stress will confer
protection against a subsequent and more severe insult (4).
This is also true for pathological stressors; for example, a brief
period of ischemia can mediate protection against a subse-
quent long-term ischemic insult (5). Protection from cell death
afforded by thermal preconditioning is mediated by HSPs as it
can be abrogated by inhibitors of HSPs such as triptolide (6).
However, although a recent study revealed that many of these
inhibitors are not as specic as initially thought (7), more spe-
cic methods, such as knockdown of HSPs, have shown their
critical role in protection from cell death (8). Interestingly,
cells exposed to a particular stress stimulus also develop
cross-tolerance to a different stress stimulus (9). For example,
thermal preconditioning can protect cells against the toxicity
of anticancer drugs (10) and the neurotoxin N-methyl-4-phe-
nylpyridine (11). The phenomenon of cross-tolerance supports
the notion that HSP induction serves as a general cytoprotec-
tive mechanism in cells.
The HSP Superfamily
HSPs function as molecular chaperones, proteins that guard
against illicit or promiscuous interactions between other
proteins. These chaperones protect the proteome from the
dangers of misfolding and aggregation by facilitating protein
folding, trafcking, complex assembly, and ubiquitination, as
well as proteasomal degradation (12). This protection is
achieved in a number of ways including through de novo pro-
tein folding, refolding of misfolded proteins, and oligomer
VC
2014 International Union of Biochemistry and Molecular Biology
Volume 66, Number 5, May 2014, Pages 327338
*Address correspondence to: Afshin Samali, Apoptosis Research Centre,
Biosciences Research Building, Corrib Village, NUI Galway, Dangan,
Galway, Ireland. Tel: 1353-9149-2440. Fax: 1353-9149-4596.
E-mail: afshin.samali@nuigalway.ie
Received 4 April 2014; Accepted 1 May 2014
DOI 10.1002/iub.1274
Published online 26 May 2014 in Wiley Online Library
(wileyonlinelibrary.com)
IUBMB Life 327
assembly. It now appears that cytoprotection is not simply a
matter of chaperoning damaged proteins but also involves
direct interference with apoptotic pathways and the mainte-
nance of cytoskeletal integrity under conditions of stress facili-
tated by the chaperoning action of HSPs (13).
HSPs are divided into families based on molecular weight:
HSPA (HSP70), HSPH (HSP110), HSPC (HSP90), DNAJ (HSP40),
HSPB (small HSPs) (14) and chaperonin families HSPD/E
(HSP60/HSP10) (15).
The human genome encodes 13 HSPA family members
including constitutively expressed and stress-inducible mem-
bers (see Table 1 for details). They all display a high degree of
sequence and domain homology. Additional characteristics
include (i) a conserved ATPase domain at the N-terminus; (ii) a
exible C-terminal peptide substrate binding domain (SBD);
and (iii) a G/P-rich C-terminal region containing an EEVD-
motif that enables the proteins to bind co-chaperones and
other HSPs. Cooperation with cofactors regulates the many
functions of HSPA proteins (16).
The HSPB family consists of 11 members, HSPB111. Some
are ubiquitously expressed, whereas others display tissue-
restricted patterns of expression (see Table 1 for details).
Furthermore, some members of stress-inducible HSPBs are
highly diverse in sequence, size, client protein specicity, and
function (17). Post-translational modications of HSPBs adds an
additional layer of regulation, which is particularly important in
stress conditions (18). The dynamic organization of HSPB
oligomers appears to be a crucial feature governing HSPB
activity. It has been demonstrated that in response to different
stress stimuli, HSPB1 alters its oligomerization status, which
may enable differential clientprotein interactions (19).
HSPD1 and HSPE1 are classed as chaperonins. HSPD1 and
HSPE1 form a complex in which HSPE1 regulates the sub-
strate binding and ATPase activity of HSPD1. A majority (60
80%) of HSPD1 and HSPE1 proteins are located in the mito-
chondria where they are involved in folding of a subset of
mitochondrial proteins (20). Roles for these proteins in the
cytoplasm are also emerging.
The HSPC family is composed of ve members designated
as HSPC15. Until recently, the literature has not differentiated
between the various family members. However, HSPC1 and
HSPC3 turned out to have non-overlapping functions (21). Spe-
cically, HSPC1 and HSPC3 show similar interaction proles
with co-chaperones but differ in their substrate interactome.
The HSPC family functions as part of a multichaperone com-
plex via association with a variety of co-chaperones. Similar to
the HSPA family, HSPC proteins have the ability to hydrolyze
ATP and to bind and modify the conformations of client pro-
teins (15,21).
The human genome encodes four HSPH family members
(HSPH14). The HSPH family is important for preventing pro-
tein aggregation. HSPH proteins are thought to primarily act
as nucleotide exchange factors for the HSPA family; however,
they have also been shown to be able to refold misfolded lucif-
erase in the absence of HSPA proteins (15).
Molecular Basis of Apoptosis
Apoptosis is a caspase-dependent form of cell death. In the
adult organism, apoptosis is responsible for the removal of
damaged, aged, or superuous cells in a manner that avoids
unwanted activation of the immune system (22). Dysregulation
of apoptosis is associated with a number of pathological proc-
esses; resistance to apoptosis can cause autoimmune disease
and cancer. Excessive apoptosis is linked with inammatory
diseases and neurodegenerative diseases (23).
The stereotypical pattern of cell demolition during apopto-
sis is mediated by a class of intracellular proteases, the cas-
pases. In mammals, caspases (cysteine-dependent aspartate-
specic proteases) are a family of 15 proteases. Seven family
members (the apoptotic caspase-2, -3, -6, -7, -8, -9, and -10)
are involved in apoptosis, whereas the remaining members
play roles in inammation and other processes. Within the
wide range of cellular targets, apoptotic caspases are key sub-
strates that mediate the demolition of the cell (24). Caspases
are present in cells as inactive dimeric proenzymes. Activation
involves two consecutive cleavage events, between the prodo-
mains and the small and large subunits, respectively, which
generates the active heterotetrameric caspase. Their prodo-
mains and activation mechanisms classify the apoptotic cas-
pases into two subgroups: initiator and executioner caspases.
Executioner caspases (caspase-3, -6, and -7) have small
prodomains, and their cleavage and activation are mediated
by other caspases. Therefore, cleavage of executioner caspases
sets in motion an irreversible chain reaction of further caspase
cleavage and activation. This cascade is initiated by initiator
caspases (caspase-2, -8, -9, and -10) that are characterized by
large prodomains that contain homotypic protein interaction
motifs facilitating proteinprotein interactions, such as caspase
activation and recruitment domains (CARD) in caspase-9 and
-2 and death effector domains (DED) in caspase-8 and
caspase-10. CARD and DED domains mediate recruitment of
capases to the so-called activation complexes whose assembly
controls the activation of the recruited initiator caspases.
There are two major pathways of initiator caspase activa-
tion. The extrinsic pathway is initiated at the cell surface by
death receptors that are members of the tumor necrosis factor
(TNF) receptor gene family. Most cellular stresses, however,
trigger the intrinsic pathway that is initiated inside cells by
mitochondrial release of proapoptotic factors.
The Extrinsic Pathway
The extrinsic pathway can be triggered by ligands of members
of the death receptor family, including TNFR, Fas/CD95,
TRAIL-R1, and TRAIL-R2 (25). The ligation of Fas or TRAIL
receptors by their specic ligands triggers their trimerization
and activation, which is followed by recruitment of the adaptor
protein Fas-Associated protein with Death Domain (FADD) at
the cytoplasmic side and assembly of the so-called death-
inducing signaling complex (DISC) that recruits procaspases-
8/-10, mediating their oligomerization and autoactivation. The
IUBMB LIFE
328 Heat Shock Proteins and Apoptosis
O
v
e
r
v
i
e
w
o
f
t
h
e
H
S
P
A
a
n
d
H
S
P
B
f
a
m
i
l
i
e
s
e
x
p
r
e
s
s
i
o
n
p
r
o

l
e
,
c
e
l
l
u
l
a
r
l
o
c
a
l
i
z
a
t
i
o
n
,
s
t
r
e
s
s
i
n
d
u
c
i
b
i
l
i
t
y
a
n
d
d
i
s
e
a
s
e
a
s
s
o
c
i
a
t
i
o
n
N
a
m
e
A
l
t
e
r
n
a
t
i
v
e
n
a
m
e
s
L
o
c
a
t
i
o
n
I
D
C
e
l
l
u
l
a
r
l
o
c
a
t
i
o
n
E
x
p
r
e
s
s
i
o
n
S
t
r
e
s
s
i
n
d
u
c
i
b
l
e
D
i
s
e
a
s
e
r
e
l
e
v
a
n
c
e
H
S
P
A
f
a
m
i
l
y
H
S
P
A
1
A
H
S
P
7
0
-
1
;
H
S
P
7
2
6
p
2
1
.
3
3
3
0
3
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
R
e
d
u
c
e
d
i
n
A
D
;
p
r
o
t
e
c
t
i
v
e
i
n
P
D
N
u
c
l
e
u
s
A
s
s
o
c
i
a
t
e
d
w
i
t
h
p
o
o
r
p
r
o
g
n
o
s
i
s
a
n
d
m
e
t
a
s
t
a
s
i
s
i
n
c
a
n
c
e
r
L
y
s
o
s
o
m
e
s
H
S
P
A
1
B
H
S
P
7
0
-
B
;
H
S
P
7
0
-
2
6
p
2
1
.
3
3
3
0
4
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
P
o
l
y
m
o
r
p
h
i
s
m
s
a
s
s
o
c
i
a
t
e
d
w
i
t
h
A
D
N
u
c
l
e
u
s
A
s
s
o
c
i
a
t
e
d
w
i
t
h
p
o
o
r
p
r
o
g
n
o
s
i
s
a
n
d
m
e
t
a
s
t
a
s
i
s
i
n
c
a
n
c
e
r
L
y
s
o
s
o
m
e
s
H
S
P
A
1
L
H
S
P
7
0
T
;
h
u
m
7
0
t
;
H
S
P
7
0
-
1
L
;
H
S
P
7
0
-
H
O
M
6
p
2
1
.
3
3
3
0
5
C
y
t
o
p
l
a
s
m
T
e
s
t
i
s
N
o
N
u
c
l
e
u
s
H
S
P
A
2
H
S
P
7
0
-
2
;
H
S
P
7
0
-
3
1
4
q
2
4
.
1
3
3
0
6
C
y
t
o
p
l
a
s
m
T
e
s
t
i
s
?
P
o
o
r
p
r
o
g
n
o
s
i
s
i
n
c
a
n
c
e
r
N
u
c
l
e
u
s
H
S
P
A
5
B
I
P
,
G
R
P
7
8
,
M
I
F
2
9
q
3
3
.
3
3
3
0
9
E
n
d
o
p
l
a
s
m
i
c
r
e
t
i
c
u
l
u
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
M
a
l
i
g
n
a
n
c
y
a
n
d
m
e
t
a
s
t
a
s
i
s
R
e
s
i
s
t
a
n
c
e
t
o
c
h
e
m
o
t
h
e
r
a
p
e
u
t
i
c
s
H
S
P
A
6
H
S
P
7
0
B
1
q
2
3
3
3
1
0
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
P
o
l
y
m
o
r
p
h
i
s
m
s
l
i
n
k
e
d
w
i
t
h
M
S
N
u
c
l
e
u
s
H
S
P
A
7
P
o
s
s
i
b
l
e
p
s
e
u
d
o
g
e
n
e
o
f
H
S
P
A
1
B
H
S
P
A
8
L
A
P
1
;
H
S
C
5
4
;
H
S
C
7
0
;
H
S
C
7
1
;
H
S
P
7
1
;
H
S
P
7
3
;
N
I
P
7
1
;
H
S
P
A
1
0
1
1
q
2
4
.
1
3
3
1
2
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
N
o
N
u
c
l
e
u
s
H
S
P
A
9
G
R
P
7
5
;
H
S
P
A
9
B
;
M
O
T
;
M
O
T
2
;
P
B
P
7
4
;
m
o
t
-
2
5
q
3
1
.
1
3
3
1
3
M
i
t
o
c
h
o
n
d
r
i
a
U
b
i
q
u
i
t
o
u
s
Y
e
s
P
o
o
r
p
r
o
g
n
o
s
i
s
i
n
c
a
n
c
e
r
T
A
B
L
E
1
Kennedy et al. 329
(
C
o
n
t
i
n
u
e
d
)
N
a
m
e
A
l
t
e
r
n
a
t
i
v
e
n
a
m
e
s
L
o
c
a
t
i
o
n
I
D
C
e
l
l
u
l
a
r
l
o
c
a
t
i
o
n
E
x
p
r
e
s
s
i
o
n
S
t
r
e
s
s
i
n
d
u
c
i
b
l
e
D
i
s
e
a
s
e
r
e
l
e
v
a
n
c
e
R
e
s
i
s
t
a
n
c
e
t
o
c
h
e
m
o
t
h
e
r
a
p
e
u
t
i
c
s
H
S
P
A
1
2
A
F
L
J
1
3
8
7
4
;
K
I
A
A
0
4
1
7
1
0
q
2
6
.
1
2
2
5
9
2
1
7
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
R
e
d
u
c
e
d
i
n
s
c
h
i
z
o
p
h
r
e
n
i
a
R
e
q
u
i
r
e
d
f
o
r
a
n
g
i
o
g
e
n
e
s
i
s
H
S
P
A
1
2
b
C
2
0
o
r
f
6
0
;
d
J
1
0
0
9
E
2
4
.
2
2
0
p
1
3
1
1
6
8
3
5
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
(
e
n
r
i
c
h
e
d
i
n
e
n
d
o
t
h
e
l
i
a
l
c
e
l
l
s
)
Y
e
s
P
r
o
t
e
c
t
i
v
e
i
n
i
s
c
h
e
m
i
a
/
r
e
p
e
r
f
u
s
i
o
n
H
S
P
A
1
3
S
T
C
H
2
1
q
1
1
6
7
8
2
M
i
c
r
o
s
o
m
e
s
U
b
i
q
u
i
t
o
u
s
?
C
a
n
c
e
r
H
S
P
B
f
a
m
i
l
y
H
S
P
B
1
C
M
T
2
F
,
H
M
N
2
B
,
H
S
.
7
6
0
6
7
,
H
S
P
2
7
,
H
S
P
2
8
,
H
s
p
2
5
,
S
R
P
2
7
7
q
1
1
.
2
3
3
3
1
5
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
C
M
T
,
d
H
M
N
N
u
c
l
e
u
s
P
e
r
i
k
a
r
y
o
n
C
a
n
c
e
r
M
e
t
a
s
t
a
s
i
s
P
r
o
t
e
c
t
i
v
e
i
n
i
s
c
h
e
m
i
a
/
r
e
p
e
r
f
u
s
i
o
n
W
i
l
l
i
a
m
s
s
y
n
d
r
o
m
e
H
S
P
B
2
M
K
B
P
;
H
S
P
2
7
;
H
s
.
7
8
8
4
6
;
L
O
H
1
1
C
R
1
K
1
1
q
2
2
-
q
2
3
3
3
1
6
C
y
t
o
s
o
l
C
a
r
d
i
a
c
,
s
k
e
l
e
t
a
l
m
u
s
c
l
e
N
o
M
y
o
p
a
t
h
y
C
y
t
o
p
l
a
s
m
I
s
c
h
e
m
i
a
N
u
c
l
e
u
s
C
a
n
c
e
r
M
i
t
o
c
h
o
n
d
r
i
a
H
S
P
B
3
H
M
N
2
C
;
D
H
M
N
2
C
;
H
S
P
L
2
7
5
q
1
1
.
2
8
9
8
8
C
y
t
o
p
l
a
s
m
C
a
r
d
i
a
c
,
s
k
e
l
e
t
a
l
m
u
s
c
l
e
N
o
d
H
M
N
N
u
c
l
e
u
s
N
e
u
r
o
p
a
t
h
y
H
S
P
B
4
C
R
Y
A
A
,
C
R
Y
A
1
,
C
T
R
C
T
9
2
1
q
2
2
.
3
1
4
0
9
C
y
t
o
p
l
a
s
m
L
e
n
s
N
o
C
a
t
a
r
a
c
t
s
N
u
c
l
e
u
s
H
S
P
B
5
C
R
Y
A
B
,
C
M
D
1
I
I
,
C
R
Y
A
2
,
C
T
P
P
2
1
1
q
2
2
.
3
-
q
2
3
.
1
1
4
1
0
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
M
y
o
p
a
t
h
y
,
n
e
u
r
o
p
a
t
h
y
N
u
c
l
e
u
s
C
a
t
a
r
a
c
t
s
C
a
n
c
e
r
M
e
t
a
s
t
a
s
i
s
P
r
o
t
e
c
t
i
v
e
i
n
i
s
c
h
e
m
i
a
/
r
e
p
e
r
f
u
s
i
o
n
T
A
B
L
E
1
IUBMB LIFE
330 Heat Shock Proteins and Apoptosis
(
C
o
n
t
i
n
u
e
d
)
N
a
m
e
A
l
t
e
r
n
a
t
i
v
e
n
a
m
e
s
L
o
c
a
t
i
o
n
I
D
C
e
l
l
u
l
a
r
l
o
c
a
t
i
o
n
E
x
p
r
e
s
s
i
o
n
S
t
r
e
s
s
i
n
d
u
c
i
b
l
e
D
i
s
e
a
s
e
r
e
l
e
v
a
n
c
e
H
S
P
B
6
H
S
P
2
0
1
9
q
1
3
.
1
2
1
2
6
3
9
3
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
Y
e
s
N
e
u
r
o
p
a
t
h
y
N
u
c
l
e
u
s
I
s
c
h
e
m
i
a
H
S
P
B
7
c
v
H
S
P
1
p
3
6
.
2
3
-
p
3
4
.
3
2
7
1
2
9
C
y
t
o
p
l
a
s
m
C
a
r
d
i
a
c
,
s
k
e
l
e
t
a
l
m
u
s
c
l
e
?
C
a
r
d
i
o
m
y
o
p
a
t
h
y
N
u
c
l
e
u
s
M
i
t
o
c
h
o
n
d
r
i
a
H
S
P
B
8
H
1
1
;
H
M
N
2
;
C
M
T
2
L
;
D
H
M
N
2
;
E
2
I
G
1
;
H
M
N
2
A
;
H
S
P
2
2
1
2
q
2
4
.
2
3
2
6
3
5
3
C
y
t
o
p
l
a
s
m
U
b
i
q
u
i
t
o
u
s
C
e
l
l
t
y
p
e
s
p
e
c
i

c
C
M
T
a
n
d
d
H
M
N
N
u
c
l
e
u
s
C
a
n
c
e
r
H
S
P
B
9
C
T
5
1
1
7
q
2
1
.
2
9
4
0
8
6
C
y
t
o
p
l
a
s
m
T
e
s
t
i
s
?
C
a
n
c
e
r
N
u
c
l
e
u
s
H
S
P
B
1
0
O
D
F
1
;
R
T
7
;
O
D
F
2
;
O
D
F
P
;
S
O
D
F
;
C
T
1
3
3
;
O
D
F
2
7
;
O
D
F
P
G
;
H
S
P
B
1
0
;
O
D
F
P
G
A
;
O
D
F
P
G
B
8
q
2
2
.
3
4
9
5
6
S
p
e
r
m
t
a
i
l
T
e
s
t
i
s
?
H
S
P
B
1
1
a
P
P
2
5
;
I
F
T
2
5
;
C
1
o
r
f
4
1
;
H
S
P
C
O
3
4
1
p
3
2
5
1
6
6
8
C
y
t
o
p
l
a
s
m
P
l
a
c
e
n
t
a
Y
e
s
C
a
n
c
e
r
N
u
c
l
e
u
s
A
b
b
r
e
v
i
a
t
i
o
n
s
:
A
D
,
A
l
z
h
e
i
m
e
r

s
d
i
s
e
a
s
e
;
P
D
,
P
a
r
k
i
n
s
o
n

s
d
i
s
e
a
s
e
;
M
S
,
m
u
l
t
i
p
l
e
s
c
l
e
r
o
s
i
s
;
C
M
T
,
C
h
a
r
c
o
t
M
a
r
i
e
T
o
o
t
h
;
d
H
M
N
,
d
i
s
t
a
l
h
e
r
e
d
i
t
a
r
y
m
u
s
c
u
l
a
r
n
e
u
r
o
p
a
t
h
y
.
a
C
o
n
t
r
o
v
e
r
s
i
a
l
H
S
P
B
m
e
m
b
e
r
a
s
i
t
d
o
e
s
n
o
t
h
a
v
e
a
c
l
a
s
s
i
c
a
l
a
-
c
r
y
s
t
a
l
l
i
n
d
o
m
a
i
n
.
T
A
B
L
E
1
Kennedy et al. 331
activated initiator caspases can directly cleave executioner
caspases, thus initiating the caspase cascade, or they cleave
the BH3-only protein BID that subsequently translocates to
mitochondria and triggers the intrinsic pathway in parallel
(described below).
The Fas receptor can also bind an alternative adaptor pro-
tein, Daxx, leading to activation of the JNK pathway. Activa-
tion of the TNF receptor allows recruitment of the adaptor
proteins (26), TNFR-Associated Death Domain and FADD.
FADD can recruit and activate procaspase-8 in a manner simi-
lar to the Fas signaling pathway. Moreover, the TNFR DISC
can prevent the activation of the prosurvival transcription fac-
tor NF-jB, thus promoting TNF-induced apoptosis (26).
Apoptosis triggered by death receptors can be impaired or
modulated by variants of the catalytically inactive caspase-8
homolog, cFLIP that can be recruited to the DISC. Important
additional regulators of DISC signaling are the inhibitor of apo-
ptosis proteins (IAPs), which mediate the ubiquitinylation of
interacting proteins at the DISC and can also impair caspase
activity (26).
The Intrinsic Pathway
Activation of the intrinsic pathway leads to mitochondrial
outer membrane permeabilization (MOMP) and release of
apoptogenic factors from the mitochondria, such as cyto-
chrome c, second mitochondrial-derived activator of caspases
(SMAC), and Omi stress-regulated endoprotease/high-tempera-
ture requirement protein A2 (HTRA2) (27). This is a crucial
event for driving caspase activation and subsequent apoptosis.
Cytochrome c binds to the apoptotic protease-activating factor
1 (Apaf-1), causing it to assemble into a large protein complex
termed the apoptosome that recruits and activates
procaspase-9 resulting in activation of downstream execu-
tioner caspases including caspases-3, -7, and -6. SMAC and
HTRA2 block X-linked inhibitor of apoptosis, a direct caspase
inhibitor of the IAP family, and thus allow for unrestrained
caspase activity (28).
MOMP is controlled by a family of proteins called the BCL-
2 family. In mammals, there are 15 core BCL-2 proteins with
varying degrees of structural similarity in short amino acid
motifs called BH (BCL-2 homology) domains designated as
BH14. The proapoptotic family members include the multido-
main members BAX and BAK (containing BH1, 2, and 3 as
well as a carboxy terminal stretch of hydrophobic amino acids
serving for membrane insertion) and the BH3-only proteins
BAD, BIM, BIK, BID, PUMA, BMF, HRK, and NOXA, whose
sequence homology to the other family members is restricted
to the BH3 domain. Antiapoptotic BCL-2 family members, BCL-
2, BCL-X
L
, MCL-1, BCL-w, BCL-2A1, and BCL-B, typically have
all four BH domains and a carboxy terminal membrane inser-
tion domain.
The dynamic interplay between these proteins regulates
MOMP. Proapoptotic multidomain family members BAX and
BAK act on the outer mitochondrial membrane and mediate
MOMP via oligomerization once they become activated by
physical interaction with BH3-only proteins. Antiapoptotic
BCL-2 members can interact with distinct BH3-only proteins,
and this interaction impairs their antiapoptotic function (29).
Thus, the BH3-only proteins trigger MOMP, and they are acti-
vated at the transcriptional or post-translational level by apo-
ptotic signaling pathways and transduce apoptotic stimuli to
the mitochondria.
Regulation of Apoptosis by HSPs
Induction of HSPs has been shown to increase resistance to
cell death induced by a number of conditions. Whether or not
increased HSP expression proves benecial or detrimental
depends on the disease in question. Some forms of cancer are
associated with increased expression of HSPs, which enhances
tumorgenicity and resistance to cell death. On the other hand,
increased expression of HSPs associated with enhanced sur-
vival of postmitotic cells such as cardiomyocytes and neurons
is considered benecial.
HSPs and Resistance to Apoptosis
It is well established that the acquisition of thermotolerance or
the overexpression of specic HSPs can attenuate apoptotic
cell death (10,30). In particular, HSPA1 and HSPB1 have con-
sistently proven to be inhibitors of apoptosis in multiple cell
types. A number of studies have provided insight into the
mechanisms involved and have shown that individual HSPs
can interact with the apoptotic pathway at several levels (31),
and these are discussed below.
HSPA Regulation of the Intrinsic Pathway
Overexpression of HSPA1 protects cells against a range of
stimuli that engage the intrinsic pathway such as hyperther-
mia (32), etoposide (33), staurosporine (34), and endoplasmic
reticulum stress (35). HSPA1 is overexpressed in many cancers
where it is associated with poor prognosis (36) and increased
metastasis, and silencing of HSPA1 has been shown to pro-
mote apoptosis in human cancer cells (37). Conversely, overex-
pression of HSPA1 is protective in neurodegenerative disease
models (38).
HSPA1 was shown to attenuate the intrinsic pathway at
three levels, upstream of, at the level of and downstream of
MOMP (see Figure 1). It has been shown that HSPA1 can act
upstream of the mitochondria by inhibiting BAX activation. In
nonapoptotic cells, BAX is present primarily in the cytosol.
Apoptotic stimuli cause a conformational change in BAX
resulting in its translocation to the mitochondria, where it
mediates MOMP. In heat-induced apoptosis models, HSPA1
can inhibit BAX activation but is unable to prevent cell death
when a constitutively membrane-targeted mutant BAX protein
is overexpressed. HSPA1 did not directly associate with BAX,
indicating that it acts upstream of BAX activation (39). Consist-
ent with this interpretation, HSPA1 overexpression prevented
the heat-induced downregulation of the antiapoptotic BCL-2
family protein MCl-1, which is an important regulator of BAX
oligomerization (40). Other studies have reported that the
IUBMB LIFE
332 Heat Shock Proteins and Apoptosis
HSPA1/Dna-J co-chaperone pair directly interacts with BAX
preventing its translocation to the mitochondria and in this
way prevents nitric oxide-induced apoptosis in macrophages
(14). Similarly, HSPA1 overexpression in HL-60 cells inhibits
Ara-C- and etoposide-induced BAX conformational change and
translocation to the mitochondria via a direct interaction (41).
Potentially, HSPA1 might bind to a protein(s) present in a com-
plex with BAX that is present in some cells but not in others.
HSPA1 was also shown to interact with the antiapoptotic BCL-
2 family member BCL2L12 preventing its proteasomal degra-
dation (42).
Several reports demonstrate that HSPA1 can interfere
with cytochrome c release. HSPA1 overexpression in macro-
phages prevents cytochrome c release from mitochondria,
thereby preventing nitric oxide-induced apoptosis (43). In
HSPA1-transfected Jurkat cells, early apoptotic events such as
mitochondrial depolarization and cytochrome c release are
inhibited following H
2
O
2
treatment (44). HSPA1 can reduce
cytochrome c release in a manner that is dependent on the
chaperoning function of HSPA1 in PEER cells subjected to heat
shock (45). However, in U937 cells, overexpression of HSPA1
failed to prevent heat-induced cytochrome c release but was
sufcient to reduce caspase activation suggesting cell type-
specic effects (46).
Downstream of the mitochondria, HSPA1 can interact
directly with components of the mitochondrial pathway and
potentially impede apoptosis. Evidence for a role downstream
of cytochrome c release has been suggested by the nding that
HSPA1 interacts with the proforms of caspases-3 and -7, but
not their activated forms, indicating that HSPA1 might act by
preventing procaspase processing (47). HSPA1 has been
reported to inhibit cytochrome c-mediated caspase activation
in vitro (8), although another study did not observe an inhibi-
tory role (32). In vitro studies have also suggested a role for
HSPA1 in the prevention of apoptosome assembly and
procaspase-9 recruitment to the apoptosome (48). However,
caution has been noted in the interpretation of these in vitro
results (49). An inhibitory role for HSPA1 even later in the
apoptotic pathway was reported downstream of caspase-3-like
proteases, where it inhibits events such as phospholipase A2
activation and changes in nuclear morphology (50).
Other Apoptosis-Regulating Mechanisms of HSPA1
There are few reports implicating HSPA1 in inhibiting the
extrinsic apoptotic pathway. In human leukemia cells, HSPA1
was shown to interact directly with TRAIL receptors, inhibiting
DISC formation and apoptosis (51). In other studies using colon
cancer cell lines, however, HSPA1 protected against TRAIL
treatment only in cell lines where the intrinsic pathway was
coactivated by TRAIL treatment. Activation of the intrinsic
pathway following activation of the extrinsic pathway can
occur in some cell types, referred to as type 2 cells. Similar
observations were made for HSPA1-mediated protection from
Fas receptor-triggered apoptosis (52), suggesting that HSPA1
mediates cytoprotection against death receptor-mediated apo-
ptosis often by impairing the intrinsic pathway.
There are several additional mechanisms, apart from the
interaction with components of the extrinsic or intrinsic apo-
ptotic pathways, by which HSPA1 can inhibit apoptosis. HSPA1
can modulate expression/activity of several prodeath stress
kinases, including apoptosis signal-regulating kinase 1 (ASK1),
JNK, and p38. HSPA1 was shown to directly bind ASK1, pre-
venting its homo-oligomerization, thereby protecting cells from
oxidative stress and death through inhibition of ASK1-
mediated cytochrome c release (53). HSPA1 can also inhibit
stress-induced JNK activation thereby preventing JNK pathway
signaling to apoptosis (32). JNK regulates the activities of both
proapoptotic and antiapoptotic members of the BCL-2 family
controlling BAX activation, and the JNK inhibitor SP600125
prevents the heat-induced depletion of MCL-1 as effectively as
HSPA1 overexpression (40).
In addition, the presence or absence of co-chaperones
such as BAG-1, BAG-3, or CHIP, which have potent antiapop-
totic properties, may be a decisive factor under specic stress
stimuli. For example, BAG-1/HSPA1 interaction favors protea-
somal degradation of certain client proteins, whereas BAG-3/
HSPA1 can protect clients such as IKKc from proteasomal deg-
radation thereby promoting the prosurvival NF-jB pathway
(54).
HSPB Regulation of the Intrinsic Pathway
HSPB1 is the best characterized member of the HSPB family
and has diverse functions, including inhibition of apoptosis,
reducing proteotoxic stress, and regulation of cytoskeleton
dynamics. Similar to HSPA1, studies revealed that HSPB1 can
impair the intrinsic pathway upstream of, at the level of, or
downstream of MOMP (see Figure 1).
Several studies demonstrated the regulation of stress
kinases such as AKT and JNK by HSPB1. These kinases modu-
late the intrinsic pathway upstream of MOMP by phosphoryl-
ating several BCL-2 family members. HSPB1 activates AKT by
promoting PI3-kinase activity, an upstream activator or AKT
(55). HSPB1, AKT, p38 MAPK, and MK2 were reported to
coexist in a signaling complex that phosphorylates AKT on
Ser-473. HSPB1 regulates AKT activation and promotes cell
survival by scaffolding MK2 to the AKT signal complex (56).
HSPB1 was reported to inhibit conformational BAX activation,
oligomerization, and translocation to the mitochondria follow-
ing metabolic stress (55). These experiments revealed no
direct interaction between HSPB1 and BAX, but suggested that
HSPB1 prevented BAX activation via PI3-kinase-mediated acti-
vation of AKT. AKT-mediated phosphorylation of BAX has
been demonstrated to suppress its translocation to mitochon-
dria (57). HSPB1-mediated AKT activation has also been
shown to cause inactivation of BH3-only protein BAD by phos-
phorylation, precluding its interaction with antiapoptotic BCL-
2 family members and promoting cell survival (55).
JNK can phosphorylate BCL-2 and BCL-X
L
thereby impair-
ing their antiapoptotic potential. Furthermore, JNK can
Kennedy et al. 333
mediate phosphorylation of BH3-only proteins BIM and BMF
preventing their sequestration by dynein and myosin motor
complexes (58). Furthermore, JNK can enhance translocation
of BAX to the mitochondria (59). Similar to HSPA1, HSPB1 has
been shown to reduce JNK activity and thereby promote cell
survival in stressed cells. Stetler et al. (60) pinpointed the
mechanism by which HSPB1 overexpression protected against
ischemic brain injury to regulation of the ASK1-JNK pathway
via a physical interaction with ASK1.
HSPB1 was found to impair the intrinsic pathway at the
level of MOMP by delaying cytochrome c release in
staurosporine-treated murine broblasts (61), and there are
several reports showing that HSPB1 blocks apoptosis down-
stream of MOMP. One study showed that a proportion of
cytosolic cytochrome c interacts with HSPB1 thereby inhibi-
ting activation of caspases and reducing the efcacy of apop-
tosome formation (62). A direct proteinprotein interaction
between HSPB1 and caspase-3 was demonstrated both in
vivo and with puried proteins in vitro. Interaction of HSPB1
with the prodomain of caspase-3 inhibits the second proteo-
lytic cleavage step necessary for full caspase-3 activation
(63).
Other Apoptosis-Regulating Mechanisms of HSPB1
HSPs, in particular HSPB1, play an important role in the regu-
lation of cytoskeleton. HSPB1 behaves as an F-actin cap-bind-
ing protein and has been shown to inhibit actin polymerization
in a phosphorylation-dependent manner as nonphosphorylat-
able HSPB1 shows reduced capacity to resist F-actin fragmen-
tation induced by H
2
O
2
and menadione (61). HSPB1-mediated
regulation of the cytoskeleton indirectly reduces activation of
the mitochondrial pathway. By binding to F-actin, HSPB1 pre-
vents cytoskeletal disruption, intracellular redistribution of
BID, cytochrome c release, and caspase-3 activation (61).
Phosphorylated HSPB1 was also found to be required for
maintenance of cell adhesion, thus suppressing apoptosis in
renal epithelial cells (64). Reactive oxygen species (ROS) can
lead to lethal oxidative damages when the antioxidant capacity
of the cell is overwhelmed. HSPB1 can protect against ROS
generated through TNFa stimulation or ROS-inducing agents
H
2
O
2
and menadione (65).
HSPB1 has been shown to bind ubiquitin and stimulate the
proteasome. Expression of HSPB1 leads to enhanced degrada-
tion of the cell cycle regulator, p27
KIP1
, while having no effect
on other cell cycle proteins such as cyclins (66). In addition, by
promoting the degradation of IjBa, HSPB1 allows NF-jB to
translocate to the nucleus promoting transcription of prosur-
vival genes (66). In contrast, a negative regulation of NF-jB by
HSPB1 has also been reported as HSPB1 was found to associ-
ate with the IKK complex, which is also involved in phospho-
rylation and ubiquitination of IjBa. In this case, however,
HSPB1 downregulated IKK signaling and thereby NF-jB acti-
vation (67). This response was specic for TNFa as the HSPB1
interaction with IKK did not change in response to interleukin-
1 treatment. Thus, similar to HSPA1, although being generally
antiapoptotic, HSPB1s effects may be stimulus and/or cell type
dependent. In addition, the phosphorylation status of HSPB1 is
an important modulator of its function as revealed in the latter
study where the phosphorylation of HSPB1 was important for
its interaction with the IKK complex. HSPB1 can protect the
eukaryotic initiation factor eIF2E from ubiquitination and sub-
sequent proteasomal degradation (68). Therefore, HSPB1
appears to exert substrate-specic effects on proteasomal deg-
radation and stability of client proteins.
Role of HSPD1 and HSPE1 in Apoptosis
The role of HSPD1 and HSPE1 in apoptosis is complex with
reports demonstrating both positive and negative regulation of
the intrinsic pathway (6971). Studies have shown that HSPD/
HSPE1 can promote caspase-3 activation. In Jurkat T cells,
HSPD1 and HSPE1 form a multiprotein complex with mito-
chondrial procaspase-3 (69,72). These data are consistent with
the chaperone function of HSPs.
In contrast, studies in cardiomyocytes show that HSPD1
and HSPE1 are cytoprotective. The use of adenoviral vectors
to overexpress both HSPD1 and HSPE1 as well as the individ-
ual overexpression of either HSPD1 or HSPE1 demonstrated
that these HSPs protect rat neonatal cardiomyocytes and
H9c2 cells against simulated ischemia, reoxygenation, and
glucose-mediated apoptosis (70,71). In general, these studies
show that the combined overexpression of HSPD1 and HSPE1
is cytoprotective and is associated with decreased cytochrome
c release, caspase activity, and DNA fragmentation (71). The
cytoprotective effects of HSPD1 were conrmed using anti-
sense oligonucleotides to knockdown the expression of this
HSP in rat cardiomyocytes (73). The cytoprotective effects of
HSPD1 are linked with regulation of BAX. Therefore, perhaps
in a manner similar to HSPA1, HSPD1 can sequester BAX in
the cytosol keeping it in an inactive conformation (74). Over-
expression of HSPD1 and HSPE1 leads to an increase in the
antiapoptotic BCL-2 and BCL-X
L
via post-transcriptional
mechanisms (74).
Other HSPs and Apoptosis
Other HSPs such as HSPB5 (aB-crystallin), HSPC (HSP90), and
HSPH1 (HSP105) have also been implicated in apoptosis regu-
lation. HSPB5 has been shown to protect cells from apoptosis
induced by staurosporine, TNFa, and Fas (65). There is evi-
dence that HSPB5 negatively regulates apoptosis by preventing
the maturation of active caspase-3 (75). Furthermore, HSPB5
can inhibit procaspase-3 processing in cytosolic extracts of
Jurkat T cells incubated with either caspase-8 or cytochrome
c/dATP, demonstrating that HSPB5 can inhibit both the
intrinsic and the extrinsic apoptotic pathways. It has also
been reported that the overexpression of BCL-2 in rabbit lens
epithelial cells sensitizes these cells to oxidative stress-induced
apoptosis through the downregulation of HSPB5 (75). Decreas-
ing BCL-2 levels through antisense RNA restored HSPB5 levels
and increased the resistance of the cells to apoptosis. Mao
et al. (76) also showed that HSPB5 is capable of binding
directly to the proapoptotic BCL-2-family members, BAX and
IUBMB LIFE
334 Heat Shock Proteins and Apoptosis
BCL-X
S
, blocking the translocation of these proteins to the
mitochondria.
HSP90 is reported to have both proapoptotic and antiapop-
totic effects depending on the stimulus. For example, in U937
monoblastoid cells, HSP90 has been shown to promote apopto-
sis induced by TNFa and cycloheximide while protecting
against UVB-induced cell death (77).
Silencing of HSP90 in PC-12 cells exposed to 6-OHDA
resulted in the inhibition of BAX activation and caspase-3
cleavage with concomitant upregulation of the antiapoptotic
protein BCL-2. This was suggested to be due to increased acti-
vation of HSF1 and a compensatory increase in HSPA1 (78).
On the other hand, HSPC1 was shown to inhibit staurosporine-
induced apoptosis in L929 and 293T cells (79). HSP90 (HSPC3)
can bind to Apaf-1 thereby impeding apoptosome formation
(79). HSP90 is able to form a complex with the antiapoptotic
RIP-1 kinase, promoting its stability, leading to increased cel-
lular survival (80). Similar to HSPB1, HSP90 (HSPC1) also reg-
ulates AKT survival signaling by preventing its inactivation.
The inhibition of HSP90 binding to AKT results in the loss of
Proposed mechanisms of heat shock protein intervention in apoptotic pathways. Both HSPA1 and HSPB1 blocked MOMP
upstream of mitochondria by inhibiting JNK and JNK activation, respectively. In addition, both HSPs were reported to directly
interfere with Bax activation and translocation, and HSPA1 was shown to prevent stress-induced MCL-1 degradation. Further-
more, HSPB1 activated AKT leading to inactivation of BH3-only proteins. HSPA1 and HSPB1 were found to delay cytochrome c
release, whereas HSPB1 bound cytochrome c and thus prevented apoptosome formation. Interference with apoptosome activ-
ity has been demonstrated for both HSPA1 and HSPB1. Finally, both HSPs inhibited the full activation of executioner procas-
pases. HSPA1 impaired death receptor signaling at the DISC, whereas HSPB1 augmented NF-jB activation by promoting the
degradation of IjB. Note that for simplicity, different death receptor signaling platforms have been unied into one. Activating
or inhibitory interactions are depicted using arrows or at lines, respectively. [Color gure can be viewed in the online issue,
which is available at wileyonlinelibrary.com.]
FIG 1
Kennedy et al. 335
AKT activity and increased sensitivity to apoptosis (81). In
addition, HSP90 associates with and stabilizes the IAP family
protein survivin. Inhibition of HSP90 chaperone function pro-
motes degradation of survivin leading to mitochondrial-
dependent apoptosis (82).
In a similar fashion to HSP90, the effects of HSPH1
(HSP105a) can be either proapoptotic or antiapoptotic. The
protective role of HSPH1 was shown in PC12 cells subjected to
various stresses, including heat shock, hydrogen peroxide, and
etoposide (83). However, a transient increase in HSPH1
expression was seen during mouse embryogenesis and, based
on experiments using mouse embryonic F9 cells, was proposed
to be associated with increased PARP cleavage, caspase-3 acti-
vation, and cytochrome c release (84).
Concluding Remarks
HSPs play a pivotal role in regulating apoptosis. HSPA1 and
HSPB1 are well-established regulators of the intrinsic pathway
by interacting with BCL-2 proteins or by modulating the activ-
ity of kinases that modulate the function of BCL-2 proteins. A
similar way of promoting cell survival has been demonstrated
for HSPC1. Furthermore, HSPB1 and HSPC1 have been sug-
gested to delay cytochrome c release in a direct fashion at the
level of mitochondria. Both HSPA1 and HSPC1 have been
shown to interfere with apoptosome formation, and all three
HSP families can interact with caspases, inhibiting their full
activation. Additional roles for HSPs in modulating DISC sig-
naling and NF-jB activation are emerging. Given the potential
contribution of alterations in HSPs expression to human dis-
ease, understanding the regulation of cell death by HSPs is a
prerequisite for novel approaches for the treatment of condi-
tions such as cancer and heart disease. In time, it is hoped
that regulation of HSPs expression, either by pharmacological
means or by gene therapy, will allow us to manipulate apopto-
tic pathways to promote or prevent cell death as required.
References
[1] Garrido, C., Gurbuxani, S., Ravagnan, L., and Kroemer, G. (2001) Heat shock
proteins: endogenous modulators of apoptotic cell death. Biochem. Biophys.
Res. Commun. 286, 433442.
[2] Morimoto, R. I. (1998) Regulation of the heat shock transcriptional response:
cross talk between a family of heat shock factors, molecular chaperones, and
negative regulators. Genes Dev. 12, 37883796.
[3] Feder, M. E., and Hofmann, G. E. (1999) Heat-shock proteins, molecular chap-
erones, and the stress response: evolutionary and ecological physiology.
Annu. Rev. Physiol. 61, 243282.
[4] Gerner, E. W., and Schneuder, M. J. (1975) Induced thermal resistance in
HeLa cells. Nature 256, 500502.
[5] Hausenloy, D. J., and Yellon, D. M. (2011) The therapeutic potential of ische-
mic conditioning: an update. Nat. Rev. Cardiol. 8, 619629.
[6] Hsu, S., Chao, C., Huang, W., Lin, M., and Cheng, B. (2013) Attenuating heat-
induced cellular autophagy, apoptosis and damage in H9c2 cardiomyocytes
by pre-inducing HSP70 with heat shock preconditioning. Int. J. Hyperthermia
29, 239247.
[7] Santagata, S., Mendillo, M. L., Tang, Y., Subramanian, A., Perley, C. C., et al.
(2013) Tight coordination of protein translation and HSF1 activation supports
the anabolic malignant state. Science 19, 341.
[8] Samali, A., Robertson, J. D., Peterson, E., Manero, F., van Zeijl, L., et al.
(2001) Hsp27 protects mitochondria of thermotolerant cells against apoptotic
stimuli. Cell Stress Chaperones 6, 4958.
[9] Li, G. C., and Hahn, G. M. (1978) Ethanol-induced tolerance to heat and to
adriamycin. Nature 274, 699701.
[10] Samali, A., and Cotter, T. G. (1996) Heat shock proteins increase resistance
to apoptosis. Exp. Cell Res. 223, 163170.
[11] Quigney, D. J., Gorman, A. M., and Samali, A. (2003) Heat shock protects
PC12 cells against MPP1 toxicity. Brain Res. 993, 133139.
[12] Balch, W. E., Morimoto, R. I., Dillin, A., and Kelly, J. W. (2008) Adapting pro-
teostasis for disease intervention. Science 319, 916919.
[13] Hartl, F. U., Bracher, A., and Hayer-Hartl, M. (2011) Molecular chaperones in
protein folding and proteostasis. Nature 475, 324332.
[14] Gotoh, T., Terada, K., Oyadomari, S., and Mori, M. (2004) hsp70-DnaJ chap-
erone pair prevents nitric oxide- and CHOP-induced apoptosis by inhibiting
translocation of bax to mitochondria. Cell Death Differ. 11, 390402.
[15] Kampinga, H., Hageman, J., Vos, M., Kubota, H., Tanguay, R., et al. (2009)
Guidelines for the nomenclature of the human heat shock proteins. Cell
Stress Chaperones 14, 105111.
[16] Daugaard, M., Rohde, M., and Jaattela, M. (2007) The heat shock protein 70
family: highly homologous proteins with overlapping and distinct functions.
FEBS Lett. 581, 37023710.
[17] Garrido, C., Paul, C., Seigneuric, R., and Kampinga, H. H. (2012) The small
heat shock proteins family: the long forgotten chaperones. Int. J. Biochem.
Cell Biol. 44, 15881592.
[18] Hatayama, T., Nishiyama, E., and Yasuda, K. (1994) Cellular localization of
high-molecular-mass heat shock proteins in murine cells. Biochem. Biophys.
Res. Commun. 200, 13671373.
[19] Paul, C., Simon, S., Gibert, B., Virot, S., Manero, F., et al. (2010) Dynamic
processes that reect anti-apoptotic strategies set up by HspB1 (Hsp27).
Exp. Cell Res. 316, 15351552.
[20] Bukau, B., and Horwich, A. L. (1998) The Hsp70 and Hsp60 chaperone
machines. Cell 92, 351366.
[21] Johnson, J. L. (2012). Evolution and function of diverse Hsp90 homologs
and cochaperone proteins. Biochim. Biophys. Acta 1823, 607613.
[22] Taylor, R. C., Cullen, S. P., and Martin, S. J. (2008) Apoptosis: controlled
demolition at the cellular level. Nat. Rev. Mol. Cell Biol. 9, 231241.
[23] Doyle, K. M., Kennedy, D., Gorman, A. M., Gupta, S., Healy, S. J. M., et al.
(2011) Unfolded proteins and endoplasmic reticulum stress in neurodege-
nerative disorders. J. Cell Mol. Med. 15, 20252039.
[24] Pop, C., and Salvesen, G. S. (2009) Human caspases: activation, specicity,
and regulation. J. Biol. Chem. 284, 2177721781.
[25] Dickens, L., Powley, I., Hughes, M., and MacFarlane, M. (2012) The com-
plexities of life and death: death receptor signalling platforms. Exp. Cell
Res. 318, 12691277.
[26] Sessler, T., Healy, S., Samali, A., and Szegezdi, E. (2013) Structural determi-
nants of DISC function: new insights into death receptor-mediated apoptosis
signalling. Pharmacol. Ther. 140, 186199.
[27] Jager, R., and Fearnhead, H. O. (2013) Mitochondrial regulation of cell-
death. In Mitochondria as Targets for Phytochemicals in Cancer Prevention
and Therapy (Chandra, D., ed.). pp. 3360, Springer, New York.
[28] Tait, S. W. G., and Green, D. R. (2010) Mitochondria and cell death: outer
membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 11, 621
632.
[29] Szegezdi, E., MacDonald, D. C., Chonghaile, N.
~
A T., Gupta, S., and Samali,
A. (2009) Bcl-2 family on guard at the ER. Am. J. Physiol. Cell Physiol. 296,
C941C953.
[30] Mosser, D. D., and Martin, L. H. (1992) Induced thermotolerance to apopto-
sis in a human T lymphocyte cell line. J. Cell Physiol. 151, 561570.
[31] Mosser, D. D., and Morimoto, R. I. (2004) Molecular chaperones and the
stress of oncogenesis. Oncogene 23, 29072918.
IUBMB LIFE
336 Heat Shock Proteins and Apoptosis
[32] Mosser, D. D., Caron, A. W., Bourget, L., Denis-Larose, C., and Massie, B.
(1997) Role of the human heat shock protein hsp70 in protection against
stress-induced apoptosis. Mol. Cell Biol. 17, 53175327.
[33] Guo, F., Rocha, K., Bali, P., Pranpat, M., Fiskus, W., et al. (2005) Abrogation
of heat shock protein 70 induction as a strategy to increase antileukemia
activity of heat shock protein 90 inhibitor 17-allylamino-demethoxy geldana-
mycin. Cancer Res. 65, 1053610544.
[34] Cheng, L., Smith, D. J., Anderson, R. L., and Nagley, P. (2011) Modulation of
cellular Hsp72 levels in undifferentiated and neuron-like SH-SY5Y cells
determines resistance to staurosporine-induced apoptosis. PLoS One. 6,
e24473.
[35] Gupta, S., Deepti, A., Deegan, S., Lisbona, F., Hetz, C., et al. (2010) HSP72
protects cells from ER stress-induced apoptosis via enhancement of IRE1
^
I6-
XBP1 signaling through a physical interaction. PLoS Biol. 8, e1000410.
[36] Thanner, F., Satterlin, M., Kapp, M., Rieger, L., Kristen, P., et al. (2003) Heat-
shock protein 70 as a prognostic marker in node-negative breast cancer.
Anticancer Res. 23, 10571062.
[37] Rohde, M., Daugaard, M., Jensen, M. H., Helin, K., Nylandsted, J., et al.
(2005) Members of the heat-shock protein 70 family promote cancer cell
growth by distinct mechanisms. Genes Dev. 19, 570582.
[38] Patel, Y. J. K., Payne Smith, M. D., de Belleroche, J., and Latchman, D. S.
(2005) Hsp27 and Hsp70 administered in combination have a potent protec-
tive effect against FALS-associated SOD1-mutant-induced cell death in
mammalian neuronal cells. Mol. Brain Res. 134, 256274.
[39] Stankiewicz, A. R., Lachapelle, G., Foo, C. P. Z., Radicioni, S. M., and
Mosser, D. D. (2005) Hsp70 inhibits heat-induced apoptosis upstream of
mitochondria by preventing bax translocation. J. Biol. Chem. 280, 38729
38739.
[40] Stankiewicz, A. R., Livingstone, A. M., Mohseni, N., and Mosser, D. D. (2009)
Regulation of heat-induced apoptosis by mcl-1 degradation and its inhibi-
tion by Hsp70. Cell Death Differ. 16, 638647.
[41] Guo, F., Sigua, C., Bali, P., George, P., Fiskus, W., et al. (2005) Mechanistic
role of heat shock protein 70 in bcr-abl mediated resistance to apoptosis in
human acute leukemia cells. Blood 105, 12461255.
[42] Yang, J., Hong, Y., Wang, W., Wu, W., Chi, Y., et al. (2009) HSP70 protects
BCL2L12 and BCL2L12A from N-terminal ubiquitination-mediated proteaso-
mal degradation. FEBS Lett. 583, 14091414.
[43] Klein, S. D., and Brane, B. (2002) Heat-shock protein 70 attenuates nitric
oxide-induced apoptosis in RAW macrophages by preventing cytochrome c
release. Biochem. J. 362, 635641.
[44] Creagh, E. M., Carmody, R. J., and Cotter, T. G. (2000) Heat shock protein
70 inhibits caspase-dependent and -independent apoptosis in Jurkat T cells.
Exp. Cell Res. 257, 5866.
[45] Mosser, D. D., Caron, A. W., Bourget, L., Meriin, A. B., Sherman, M. Y.,
et al. (2000) The chaperone function of hsp70 is required for protection
against stress-induced apoptosis. Mol. Cell Biol. 20, 71467159.
[46] Li, C., Lee, J., Ko, Y., Kim, J., and Seo, J. (2000) Heat shock protein 70 inhib-
its apoptosis downstream of cytochrome c release and upstream of
caspase-3 activation. J. Biol. Chem. 275, 2566525671.
[47] Komarova, E. Y., Afanasyeva, E. A., Bulatova, M. M., Cheetham, M. E.,
Margulis, B. A., et al. (2004) Downstream caspases are novel targets for the
antiapoptotic activity of the molecular chaperone Hsp70. Cell Stress Chaper-
ones 9, 265275.
[48] Beere, H. M., Wolf, B. B., Cain, K., Mosser, D. D., Mahboubi, A., et al. (2000)
Heat-shock protein 70 inhibits apoptosis by preventing recruitment of
procaspase-9 to the apaf-1 apoptosome. Nat. Cell Biol. 2, 469475.
[49] Steel, R., Doherty, J. P., Buzzard, K., Clemons, N., Hawkins, C. J., et al.
(2004) Hsp72 inhibits apoptosis upstream of the mitochondria and not
through interactions with apaf-1. J. Biol. Chem. 279, 5149051499.
[50] Jaattelaa, M., Wissing, D., Kokholm, K., Kallunki, T., and Egeblad, M. (1998)
Hsp70 exerts its anti-apoptotic function downstream of caspase-3-like pro-
teases. EMBO J. 17, 61246134.
[51] Ozoren, N., and El-Deiry, W. (2002) Heat shock protects HCT116 and H460
cells from TRAIL-induced apoptosis. Exp. Cell Res. 281, 175181.
[52] Clemons, N. J., Buzzard, K., Steel, R., and Anderson, R. L. (2005) Hsp72
inhibits fas-mediated apoptosis upstream of the mitochondria in type II
cells. J. Biol. Chem. 280, 90059012.
[53] Park, H., Cho, S., Kim, C. K., Hwang, H. S., Noh, K. T., et al. (2002) Heat
shock protein Hsp72 is a negative regulator of apoptosis signal-regulating
kinase 1. Mol. Cell Biol. 22, 77217730.
[54] Ammirante, M., Rosati, A., Arra, C., Basile, A., Falco, A., et al. (2010) IKKc
protein is a target of BAG3 regulatory activity in human tumor growth.
Proc. Natl. Acad. Sci. USA 107, 74977502.
[55] Havasi, A., Li, Z., Wang, Z., Martin, J. L., Botla, V., et al. (2008) Hsp27 inhib-
its bax activation and apoptosis via a phosphatidylinositol 3-kinase-
dependent mechanism. J. Biol. Chem. 283, 1230512313.
[56] Wu, R., Kausar, H., Johnson, P., Montoya-Durango, D., Merchant, M., et al.
(2007) Hsp27 regulates akt activation and polymorphonuclear leukocyte
apoptosis by scaffolding MK2 to akt signal complex. J. Biol. Chem. 282,
2159821608.
[57] Tsuruta, F., Masuyama, N., and Gotoh, Y. (2002) The phosphatidylinositol 3-
kinase (PI3K)-akt pathway suppresses bax translocation to mitochondria. J.
Biol. Chem. 277, 1404014047.
[58] Lei, K., and Davis, R. J. (2003) JNK phosphorylation of bim-related members
of the Bcl2 family induces bax-dependent apoptosis. Proc. Natl. Acad Sci.
USA 100, 24322437.
[59] Tsuruta, F., Sunayama, J., Mori, Y., Hattori, S., Shimizu, S., et al. (2004) JNK
promotes bax translocation to mitochondria through phosphorylation of 14-
3-3 proteins. EMBO J. 23, 18891899.
[60] Stetler, R. A., Cao, G., Gao, Y., Zhang, F., Wang, S., et al. (2008) Hsp27 pro-
tects against ischemic brain injury via attenuation of a novel stress-
response cascade upstream of mitochondrial cell death signaling. J. Neuro-
sci. 28, 1303813055.
[61] Paul, C., Manero, F., Gonin, S., Kretz-Remy, C., Virot, S., et al. (2002) Hsp27
as a negative regulator of cytochrome c release. Mol. Cell Biol. 22, 816834.
[62] Concannon, C. G., Orrenius, S., and Samali, A. (2004) Hsp27 inhibits cyto-
chrome c-mediated caspase activation by sequestering both pro-caspase-3
and cytochrome c. Gene Exp. 9, 195201.
[63] Voss, O. H., Batra, S., Kolattukudy, S. J., Gonzalez-Mejia, M., Smith, J. B.,
et al. (2007) Binding of caspase-3 prodomain to heat shock protein 27 regu-
lates monocyte apoptosis by inhibiting caspase-3 proteolytic activation. J.
Biol. Chem. 282, 2508825099.
[64] de Graauw, M., Tijdens, I., Cramer, R., Corless, S., Timms, J. F., et al. (2005)
Heat shock protein 27 is the major differentially phosphorylated protein
involved in renal epithelial cellular stress response and controls focal adhe-
sion organization and apoptosis. J. Biol. Chem. 280, 2988529898.
[65] Mehlen, P., Preville, X., Chareyron, P., Briolay, J., Klemenz, R., et al. (1995)
Constitutive expression of human hsp27, drosophila hsp27, or human a B-
crystallin confers resistance to TNF- and oxidative stress-induced cytotoxic-
ity in stably transfected murine L929 broblasts. J. Immunol. 154, 363374.
[66] Parcellier, A., Schmitt, E., Gurbuxani, S., Seigneurin-Berny, D., Pance, A.,
et al. (2003) HSP27 is a ubiquitin-binding protein involved in IKKb proteaso-
mal degradation. Mol. Cell Biol. 23, 57905802.
[67] Park, K., Gaynor, R. B., and Kwak, Y. T. (2003) Heat shock protein 27 associ-
ation with the IKKb kinase complex regulates tumor necrosis factor a-
induced NF-jB activation. J. Biol. Chem. 278, 3527235278.
[68] Andrieu, C., Taieb, D., Baylot, V., Ettinger, S., Soubeyran, P., et al. (2010)
Heat shock protein 27 confers resistance to androgen ablation and chemo-
therapy in prostate cancer cells through eIF4E. Oncogene 29, 18831896.
[69] Samali, A., Cai, J., Zhivotovsky, B., Jones, D. P., and Orrenius, S. (1999)
Presence of a pre-apoptotic complex of pro-caspase-3, Hsp60 and Hsp10 in
the mitochondrial fraction of Jurkat cells. 18, 20402048.
[70] Lau, S., Patnaik, N., Sayen, M. R., and Mestril, R. (1997) Simultaneous over-
expression of two stress proteins in rat cardiomyocytes and myogenic cells
confers protection against ischemia-induced injury. Circulation 96, 2287
2294.
[71] Lin, K. M., Lin, B., Lian, I. Y., Mestril, R., Schefer, I. E., et al. (2001) Com-
bined and individual mitochondrial HSP60 and HSP10 expression in cardiac
myocytes protects mitochondrial function and prevents apoptotic cell
Kennedy et al. 337
deaths induced by simulated ischemia-reoxygenation. Circulation 103,
17871792.
[72] Xanthoudakis, S., Roy, S., Rasper, D., Hennessey, T., Aubin, Y., et al. (1999)
Hsp60 accelerates the maturation of pro-caspase-3 by upstream activator
proteases during apoptosis. EMBO J. 18, 20492056.
[73] Kirchhoff, S. R., Gupta, S., and Knowlton, A. A. (2002) Cytosolic heat shock
protein 60, apoptosis, and myocardial injury. Circulation 105, 28992904.
[74] Gupta, S., and Knowlton, A. A. (2005) HSP60, bax, apoptosis and the heart.
J. Cell Mol. Med. 9, 5158.
[75] Mao, Y., Xiang, H., Wang, J., Korsmeyer, S., Reddan, J., et al. (2001) Human
bcl-2 gene attenuates the ability of rabbit lens epithelial cells against H
2
O
2
-
induced apoptosis through down-regulation of the
^
I6B-crystallin gene. J.
Biol. Chem. 276, 4343543445.
[76] Mao, Y., Liu, J., Xiang, H., and Li, D. W. (2004) Human aA- and aB-
crystallins bind to bax and bcl-XS to sequester their translocation during
staurosporine-induced apoptosis. Cell Death Differ. 11, 512526.
[77] Galea-Lauri, J., Richardson, A. J., Latchman, D. S., and Katz, D. R. (1996)
Increased heat shock protein 90 (hsp90) expression leads to increased apo-
ptosis in the monoblastoid cell line U937 following induction with TNF-a
and cycloheximide: a possible role in immunopathology. J. Immunol. 157,
41094118.
[78] Alani, B., Salehi, R., Sadeghi, P., Zare, M., Khodagholi, F., et al. (2014)
Silencing of Hsp90 chaperone expression protects against 6-
hydroxydopamine toxicity in PC12 cells. J. Mol. Neurosci. 52, 392402.
[79] Pandey, P., Saleh, A., Nakazawa, A., Kumar, S., Srinivasula, S. M., et al.
(2000) Negative regulation of cytochrome c mediated oligomerization of
Apaf1 and activation of procaspase-9 by heat shock protein 90. EMBO J. 19,
43104322.
[80] Lewis, J., Devin, A., Miller, A., Lin, Y., Rodriguez, Y., et al. (2000) Disruption
of Hsp90 function results in degradation of the death domain kinase,
receptor-interacting protein (RIP), and blockage of tumor necrosis factor-
induced nuclear factor-
^
I

B activation. J. Biol. Chem. 275, 1051910526.


[81] Sato, S., Fujita, N., and Tsuruo, T. (2000) Modulation of akt kinase activity
by binding to Hsp90. Proc. Natl. Acad. Sci. USA 97, 1083210837.
[82] Fortugno, P., Beltrami, E., Plescia, J., Fontana, J., Pradhan, D., et al. (2003)
Regulation of survivin function by Hsp90. Proc. Natl. Acad. Sci. USA 100,
1379113796.
[83] Hatayama, T., Yamagishi, N., Minobe, E., and Sakai, K. (2001) Role of
hsp105 in protection against stress-induced apoptosis in neuronal PC12
cells. Biochem. Biophys. Res. Commun. 288, 528534.
[84] Yamagishi, N., Saito, Y., Ishihara, K., and Hatayama, T. (2002) Enhancement
of oxidative stress-induced apoptosis by Hsp105a in mouse embryonal F9
cells. Eur. J. Biochem. 269, 41434151.
IUBMB LIFE
338 Heat Shock Proteins and Apoptosis

You might also like